51
|
Treelet transform analysis to identify clusters of systemic inflammatory variance in a population with moderate-to-severe traumatic brain injury. Brain Behav Immun 2021; 95:45-60. [PMID: 33524553 PMCID: PMC9004489 DOI: 10.1016/j.bbi.2021.01.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/20/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Inflammatory cascades following traumatic brain injury (TBI) can have both beneficial and detrimental effects on recovery. Single biomarker studies do not adequately reflect the major arms of immunity and their relationships to long-term outcomes. Thus, we applied treelet transform (TT) analysis to identify clusters of interrelated inflammatory markers reflecting major components of systemic immune function for which substantial variation exists among individuals with moderate-to-severe TBI. METHODS Serial blood samples from 221 adults with moderate-to-severe TBI were collected over 1-6 months post-injury (n = 607 samples). Samples were assayed for 33 inflammatory markers using Millipore multiplex technology. TT was applied to standardized mean biomarker values generated to identify latent patterns of correlated markers. Treelet clusters (TC) were characterized by biomarkers related to adaptive immunity (TC1), innate immunity (TC2), soluble molecules (TC3), allergy immunity (TC4), and chemokines (TC5). For each TC, a score was generated as the linear combination of standardized biomarker concentrations and cluster load for each individual in the cohort. Ordinal logistic or linear regression was used to test associations between TC scores and 6- and 12-month Glasgow Outcome Scale (GOS), Disability Rating Scale (DRS), and covariates. RESULTS When adjusting for clinical covariates, TC5 was significantly associated with 6-month GOS (odds ratio, OR = 1.44; p-value, p = 0.025) and 6-month DRS scores (OR = 1.46; p = 0.013). TC5 relationships were attenuated when including all TC scores in the model (GOS: OR = 1.29, p = 0.163; DRS: OR = 1.33, p = 0.100). When adjusting for all TC scores and covariates, only TC3 was associated with 6- and 12-month GOS (OR = 1.32, p = 0.041; OR = 1.39, p = 0.002) and also 6- and 12-month DRS (OR = 1.38, p = 0.016; OR = 1.58, p = 0.0002). When applying TT to inflammation markers significantly associated with 6-month GOS, multivariate modeling confirmed that TC3 remained significantly associated with GOS. Biomarker cluster membership remained consistent between the GOS-specific dendrogram and overall dendrogram. CONCLUSIONS TT effectively characterized chronic, systemic immunity among a cohort of individuals with moderate-to-severe TBI. We posit that chronic chemokine levels are effector molecules propagating cellular immune dysfunction, while chronic soluble receptors are inflammatory damage readouts perpetuated, in part, by persistent dysfunctional cellular immunity to impact neuro-recovery.
Collapse
|
52
|
Gozt A, Hellewell S, Ward PGD, Bynevelt M, Fitzgerald M. Emerging Applications for Quantitative Susceptibility Mapping in the Detection of Traumatic Brain Injury Pathology. Neuroscience 2021; 467:218-236. [PMID: 34087394 DOI: 10.1016/j.neuroscience.2021.05.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a common but heterogeneous injury underpinned by numerous complex and interrelated pathophysiological mechanisms. An essential trace element, iron is abundant within the brain and involved in many fundamental neurobiological processes, including oxygen transportation, oxidative phosphorylation, myelin production and maintenance, as well as neurotransmitter synthesis and metabolism. Excessive levels of iron are neurotoxic and thus iron homeostasis is tightly regulated in the brain, however, many details about the mechanisms by which this is achieved are yet to be elucidated. A key mediator of oxidative stress, mitochondrial dysfunction and neuroinflammatory response, iron dysregulation is an important contributor to secondary injury in TBI. Advances in neuroimaging that leverage magnetic susceptibility properties have enabled increasingly comprehensive investigations into the distribution and behaviour of iron in the brain amongst healthy individuals as well as disease states such as TBI. Quantitative Susceptibility Mapping (QSM) is an advanced neuroimaging technique that promises quantitative estimation of local magnetic susceptibility at the voxel level. In this review, we provide an overview of brain iron and its homeostasis, describe recent advances enabling applications of QSM within the context of TBI and summarise the current state of the literature. Although limited, the emergent research suggests that QSM is a promising neuroimaging technique that can be used to investigate a host of pathophysiological changes that are associated with TBI.
Collapse
Affiliation(s)
- Aleksandra Gozt
- Curtin University, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Bentley, WA Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA Australia
| | - Sarah Hellewell
- Curtin University, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Bentley, WA Australia
| | - Phillip G D Ward
- Australian Research Council Centre of Excellence for Integrative Brain Function, VIC Australia; Turner Institute for Brain and Mental Health, Monash University, VIC Australia
| | - Michael Bynevelt
- Neurological Intervention and Imaging Service of Western Australia, Sir Charles Gairdner Hospital, Nedlands, WA Australia
| | - Melinda Fitzgerald
- Curtin University, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Bentley, WA Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA Australia.
| |
Collapse
|
53
|
Karelina K, Schneiderman K, Shah S, Fitzgerald J, Cruz RV, Oliverio R, Whitehead B, Yang J, Weil ZM. Moderate Intensity Treadmill Exercise Increases Survival of Newborn Hippocampal Neurons and Improves Neurobehavioral Outcomes after Traumatic Brain Injury. J Neurotrauma 2021; 38:1858-1869. [PMID: 33470170 PMCID: PMC8219196 DOI: 10.1089/neu.2020.7389] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Physician-prescribed rest after traumatic brain injury (TBI) is both commonplace and an increasingly scrutinized approach to TBI treatment. Although this practice remains a standard of patient care for TBI, research of patient outcomes reveals little to no benefit of prescribed rest after TBI, and in some cases prolonged rest has been shown to interfere with patient well-being. In direct contrast to the clinical advice regarding physical activity after TBI, animal models of brain injury consistently indicate that exercise is neuroprotective and promotes recovery. Here, we assessed the effect of low and moderate intensity treadmill exercise on functional outcome and hippocampal neural proliferation after brain injury. Using the controlled cortical impact (CCI) mouse model of TBI, we show that 10 days of moderate intensity treadmill exercise initiated after CCI reduces anxiety-like behavior, improves hippocampus-dependent spatial memory, and promotes hippocampal proliferation and newborn neuronal survival. Pathophysiological measures including lesion volume and axon degeneration were not altered by exercise. Taken together, these data reveal that carefully titrated physical activity may be a safe and effective approach to promoting recovery after brain injury.
Collapse
Affiliation(s)
- Kate Karelina
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Katarina Schneiderman
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Sarthak Shah
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Julie Fitzgerald
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Ruth Velazquez Cruz
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Robin Oliverio
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Bailey Whitehead
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Jingzhen Yang
- Nationwide Children's Hospital, Center for Injury Research and Policy, Columbus, Ohio, USA
| | - Zachary M. Weil
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
54
|
Cell Proliferation in the Piriform Cortex of Rats with Motor Cortex Ablation Treated with Growth Hormone and Rehabilitation. Int J Mol Sci 2021; 22:ijms22115440. [PMID: 34064044 PMCID: PMC8196768 DOI: 10.3390/ijms22115440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/17/2022] Open
Abstract
Traumatic brain injury represents one of the main health problems in developed countries. Growth hormone (GH) and rehabilitation have been claimed to significantly contribute to the recovery of lost motor function after acquired brain injury, but the mechanisms by which this occurs are not well understood. In this work, we have investigated cell proliferation in the piriform cortex (PC) of adult rats with ablation of the frontal motor cortex treated with GH and rehabilitation, in order to evaluate if this region of the brain, related to the sense of smell, could be involved in benefits of GH treatment. Male rats were either ablated the frontal motor cortex in the dominant hemisphere or sham-operated and treated with GH or vehicle at 35 days post-injury (dpi) for five days. At 36 dpi, all rats received daily injections of bromodeoxyuridine (BrdU) for four days. We assessed motor function through the paw-reaching-for-food task. GH treatment and rehabilitation at 35 dpi significantly improved the motor deficit caused by the injury and promoted an increase of cell proliferation in the PC ipsilateral to the injury, which could be involved in the improvement observed. Cortical ablation promoted a greater number of BrdU+ cells in the piriform cortex that was maintained long-term, which could be involved in the compensatory mechanisms of the brain after injury.
Collapse
|
55
|
Zimering MB, Delic V, Citron BA. Gene Expression Changes in a Model Neuron Cell Line Exposed to Autoantibodies from Patients with Traumatic Brain Injury and/or Type 2 Diabetes. Mol Neurobiol 2021; 58:4365-4375. [PMID: 34013450 PMCID: PMC8487420 DOI: 10.1007/s12035-021-02428-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/11/2021] [Indexed: 11/24/2022]
Abstract
Traumatic brain injury and adult type 2 diabetes mellitus are each associated with the late occurrence of accelerated cognitive decline and Parkinson’s disease through unknown mechanisms. Previously, we reported increased circulating agonist autoantibodies targeting the 5-hydroxytryptamine 2A receptor in plasma from subsets of Parkinson’s disease, dementia, and diabetic patients suffering with microvascular complications. Here, we use a model neuron, mouse neuroblastoma (N2A) cell line, to test messenger RNA expression changes following brief exposure to traumatic brain injury and/or type 2 diabetes mellitus plasma harboring agonist 5-hydroxytryptamine 2A receptor autoantibodies. We now report involvement of the mitochondrial dysfunction pathway and Parkinson’s disease pathways in autoantibody-induced gene expression changes occurring in neuroblastoma cells. Functional gene categories upregulated significantly included cell death, cytoskeleton-microtubule function, actin polymerization or depolymerization, regulation of cell oxidative stress, mitochondrial function, immune function, protein metabolism, and vesicle function. Gene categories significantly downregulated included microtubule function, cell adhesion, neurotransmitter release, dopamine metabolism synaptic plasticity, maintenance of neuronal differentiation, mitochondrial function, and cell signaling. Taken together, these results suggest that agonist 5-hydroxytryptamine receptor autoantibodies (which increase in Parkinson’s disease and other forms of neurodegeneration) mediate a coordinating program of gene expression changes in a model neuron which predispose to neuro-apoptosis and are linked to human neurodegenerative diseases pathways.
Collapse
Affiliation(s)
- Mark B Zimering
- Endocrine and Diabetes Section, Medical Service, VA New Jersey Healthcare System, 385 Tremont Ave, East Orange, NJ, 07018, USA. .,Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| | - Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA.,Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|
56
|
Du J, Zhang A, Li J, Liu X, Wu S, Wang B, Wang Y, Jia H. Doxorubicin-Induced Cognitive Impairment: The Mechanistic Insights. Front Oncol 2021; 11:673340. [PMID: 34055643 PMCID: PMC8158153 DOI: 10.3389/fonc.2021.673340] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Chemotherapy can significantly prolong the survival of patients with breast cancer; Nevertheless, the majority of patients receiving chemotherapy such as doxorubicin may have cognitive deficits that manifest as impairments in learning, reasoning, attention, and memory. The phenomenon of chemotherapy-induced cognitive decline is termed as chemotherapy-related cognitive impairment (CRCI) or chemo-brain. Doxorubicin (DOX), a commonly used drug in adjuvant chemotherapy for patients with breast cancer, has been reported to induce chemo-brain through a variety of mechanisms including DNA damage, oxidative stress, inflammation, dysregulation of apoptosis and autophagy, changes in neurotransmitter levels, mitochondrial dysfunction, glial cell interactions, neurogenesis inhibition, and epigenetic factors. These mechanisms do not operate independently but are inter-related, coordinately contributing to the development of chemo-brain. Here we review the relationships of these mechanisms and pathways in attempt to provide mechanistic insights into the doxorubicin-induced cognitive impairment.
Collapse
Affiliation(s)
- Jiajia Du
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Aoxue Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Xin Liu
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuai Wu
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Bin Wang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
57
|
Nessel I, Michael-Titus AT. Lipid profiling of brain tissue and blood after traumatic brain injury. Semin Cell Dev Biol 2021; 112:145-156. [DOI: 10.1016/j.semcdb.2020.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 11/15/2022]
|
58
|
Rakib F, Al-Saad K, Ahmed T, Ullah E, Barreto GE, Md Ashraf G, Ali MHM. Biomolecular alterations in acute traumatic brain injury (TBI) using Fourier transform infrared (FTIR) imaging spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 248:119189. [PMID: 33277210 DOI: 10.1016/j.saa.2020.119189] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 10/27/2020] [Accepted: 11/01/2020] [Indexed: 06/12/2023]
Abstract
Acute injury is one of the substantial stage post-traumatic brain injury (TBI) occurring at the moment of impact. Decreased metabolism, unregulated cerebral blood flow and direct tissue damage are triggered by acute injury. Understating the biochemical alterations associated with acute TBI is critical for brain plasticity and recovery. The objective of this study was to investigate the biochemical and molecular changes in hippocampus, corpus callosum and thalamus brain regions post-acute TBI in rats. Fourier Transform Infrared (FTIR) imaging spectroscopy were used to collect chemical images from control and 3 hrs post-TBI (Marmarou model was used for the TBI induction) rat brains and adjacent sections were treated by hematoxylin and eosin (H&E) staining to correlate with the disruption in tissue morphology and injured brain biochemistry. Our results revealed that the total lipid and total protein content decreased significantly in the hippocampus, corpus callosum and thalamus after brain injury. Reduction in lipid acyl chains (-CH2) associated with an increase in methyl (-CH3) and unsaturated lipids olefin = CH concentrations is observed. Furthermore, there is a decrease in the lipid order (disorder), which leads to an increase in acyl chain fluidity in injured rats. The results suggest acute TBI damages brain tissues mechanically rather than chemical alterations. This will help in assessing successful therapeutic strategy in order to mitigate tissue damage in acute TBI period.
Collapse
Affiliation(s)
- Fazle Rakib
- Department of Chemistry and Earth Sciences, Qatar University, Doha, Qatar
| | - Khalid Al-Saad
- Department of Chemistry and Earth Sciences, Qatar University, Doha, Qatar
| | - Tariq Ahmed
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Ehsan Ullah
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Limerick, Ireland
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Mohamed H M Ali
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
59
|
Wang J, Hou Y, Zhang L, Liu M, Zhao J, Zhang Z, Ma Y, Hou W. Estrogen Attenuates Traumatic Brain Injury by Inhibiting the Activation of Microglia and Astrocyte-Mediated Neuroinflammatory Responses. Mol Neurobiol 2021; 58:1052-1061. [PMID: 33085047 DOI: 10.1007/s12035-020-02171-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022]
Abstract
Traumatic brain injury (TBI), which leads to high mortality and morbidity, is a prominent public health problem worldwide. Neuroinflammation involving microglia and astrocyte activation has been demonstrated to play critical role in the secondary injury induced by TBI. A1 astrocytes, which are induced by activated microglia, can directly kill neurons by secreting neurotoxic complement C3. Estrogen has been proved to possess neuroprotective effects, but the effect and underlying mechanism of estrogen on TBI-induced neuroinflammatory injury remain largely unclear. In this study, we constructed an adult male mouse model of TBI and immediately after injury treated the mice with 17β-estradiol (E2) (100 μg/kg, once every day via intraperitoneal injection) for 3 days. We found that E2 treatment significantly alleviated TBI-induced neurological deficits, neuronal injuries, and brain edema and significantly inhibited Iba1 and GFAP expression, which are markers of microglia and astrocyte activation, respectively. E2 treatment also significantly inhibited TLR4 and NF-κB protein expression, and significantly reduced the expression of the proinflammatory factors IL-1β, IL-6, and TNF-α. Moreover, E2 treatment significantly decreased the number of complement C3d/GFAP-positive cells and complement C3d protein expression. Taking these results together, we concluded that E2 treatment dramatically alleviates TBI neuroinflammatory injury by inhibiting TLR4/NF-κB pathway-mediated microglia and astrocyte activation and neuroinflammation and reducing A1-phenotype neurotoxic astrocyte activation. Our findings indicate that E2 treatment may be a potential therapy strategy for TBI-induced neuroinflammation injury.
Collapse
Affiliation(s)
- Jin Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, China
| | - Yushu Hou
- Department of Anesthesiology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710001, China
| | - Lixia Zhang
- Department of Burn and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Min Liu
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, No. 28, Fuxing Road, Beijing, 100853, China
| | - Jianshuai Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, China
| | - Zhen Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, China
| | - Yulong Ma
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, No. 28, Fuxing Road, Beijing, 100853, China.
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
60
|
Smith NIJ, Gilmour S, Prescott-Mayling L, Hogarth L, Corrigan JD, Williams WH. A pilot study of brain injury in police officers: A source of mental health problems? J Psychiatr Ment Health Nurs 2021; 28:43-55. [PMID: 32662181 DOI: 10.1111/jpm.12676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/31/2020] [Accepted: 07/08/2020] [Indexed: 11/29/2022]
Abstract
WHAT IS KNOWN ON THE SUBJECT?: Traumatic brain injury (TBI) has been linked to poor outcomes in terms of mental health, specifically, PTSD, depression and alcohol abuse. A lack of research evidence exists relevant to exploring the presence and implications of TBI in the police in the UK and globally, despite the elevated risk of physical and emotional trauma specific to policing. WHAT DOES THE PAPER ADD TO EXISTING KNOWLEDGE?: The rate of traumatic brain injury is highly prevalent in a small sample of police officers. Traumatic brain injury is a major source of post-concussion symptoms (physical, cognitive and emotional deficits) in police officers, which, in general, are associated with greater mental health difficulties and drinking alcohol to cope. WHAT ARE THE IMPLICATIONS FOR PRACTICE?: Traditional mental health treatments should be supplemented with elements of concussion care to address any cognitive, emotional and physical issues due to head injury. Interventions should be made more accessible to those suffering from a mild brain injury. This can be done through regular reminders of appointments, pictograms and by providing a concrete follow-up. ABSTRACT: Introduction Police officers have a high risk of injury through assaults, road traffic incidents and attending domestic calls, with many officers developing post-traumatic stress disorder (PTSD) as a consequence. Traumatic brain injury (TBI) is a common injury in populations involved in conflict and has been extensively linked to mental health difficulties. However, current research has not explored the frequency and sequelae of TBI in police populations, despite the elevated risk of physical and emotional trauma specific to policing. Aim To explore self-reported TBI, PTSD, post-concussion symptoms, depression and drinking to cope in a small sample of UK police, to determine the frequency of these conditions and their relationships. Method Measures of TBI, mental health, and drinking alcohol to cope were administered to 54 police officers from a Midshire Police Constabulary. Results Mild TBI with loss of consciousness was reported by 38.9% of the sample. TBI was associated with increased post-concussion symptoms (PCS). PCS were associated with greater severity of PTSD, depression and drinking to cope. Discussion Exploring TBI in the police could identify a major factor contributing towards ongoing mental health difficulties in a population where, based on previous research, the implications of TBI should not be overlooked, highlighting the need for further research in this area. Implications for Practice This research spans to identify the importance of routine assessment and increasing awareness within mental health services. Mental health treatments should be made amenable to a population with potential memory, planning and impulse control deficits. Further work in mental health services is needed to understand the level of ongoing issues that are due to post-concussion symptoms and those that are due to other mental health difficulties, such as PTSD, thereby educating patients on the association between TBI and emotional difficulties. A graduated return-to-work plan should be developed to enable a safe transition back to work, whilst managing any ongoing symptoms.
Collapse
Affiliation(s)
- Nicholas I J Smith
- School of Psychology, Washington Singer Laboratories, University of Exeter, Exeter, UK
| | | | | | - Lee Hogarth
- School of Psychology, Washington Singer Laboratories, University of Exeter, Exeter, UK
| | - John D Corrigan
- Department of Physical Medicine & Rehabilitation, The Ohio State University, Columbus, OH, USA
| | - W Huw Williams
- School of Psychology, Washington Singer Laboratories, University of Exeter, Exeter, UK
| |
Collapse
|
61
|
Huntemer-Silveira A, Patil N, Brickner MA, Parr AM. Strategies for Oligodendrocyte and Myelin Repair in Traumatic CNS Injury. Front Cell Neurosci 2021; 14:619707. [PMID: 33505250 PMCID: PMC7829188 DOI: 10.3389/fncel.2020.619707] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/07/2020] [Indexed: 12/18/2022] Open
Abstract
A major consequence of traumatic brain and spinal cord injury is the loss of the myelin sheath, a cholesterol-rich layer of insulation that wraps around axons of the nervous system. In the central nervous system (CNS), myelin is produced and maintained by oligodendrocytes. Damage to the CNS may result in oligodendrocyte cell death and subsequent loss of myelin, which can have serious consequences for functional recovery. Demyelination impairs neuronal function by decelerating signal transmission along the axon and has been implicated in many neurodegenerative diseases. After a traumatic injury, mechanisms of endogenous remyelination in the CNS are limited and often fail, for reasons that remain poorly understood. One area of research focuses on enhancing this endogenous response. Existing techniques include the use of small molecules, RNA interference (RNAi), and monoclonal antibodies that target specific signaling components of myelination for recovery. Cell-based replacement strategies geared towards replenishing oligodendrocytes and their progenitors have been utilized by several groups in the last decade as well. In this review article, we discuss the effects of traumatic injury on oligodendrocytes in the CNS, the lack of endogenous remyelination, translational studies in rodent models promoting remyelination, and finally human clinical studies on remyelination in the CNS after injury.
Collapse
Affiliation(s)
| | - Nandadevi Patil
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Megan A. Brickner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Ann M. Parr
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
62
|
Montivero AJ, Ghersi MS, Catalán-Figueroa J, Formica ML, Camacho N, Culasso AF, Hereñú CB, Palma SD, Pérez MF. Beyond Acute Traumatic Brain Injury: Molecular Implications of Associated Neuroinflammation in Higher-Order Cognitive Processes. PSYCHIATRY AND NEUROSCIENCE UPDATE 2021:237-259. [DOI: 10.1007/978-3-030-61721-9_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
63
|
Kövesdi E, Szabó-Meleg E, Abrahám IM. The Role of Estradiol in Traumatic Brain Injury: Mechanism and Treatment Potential. Int J Mol Sci 2020; 22:E11. [PMID: 33374952 PMCID: PMC7792596 DOI: 10.3390/ijms22010011] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023] Open
Abstract
Patients surviving traumatic brain injury (TBI) face numerous neurological and neuropsychological problems significantly affecting their quality of life. Extensive studies over the past decades have investigated pharmacological treatment options in different animal models, targeting various pathological consequences of TBI. Sex and gender are known to influence the outcome of TBI in animal models and in patients, respectively. Apart from its well-known effects on reproduction, 17β-estradiol (E2) has a neuroprotective role in brain injury. Hence, in this review, we focus on the effect of E2 in TBI in humans and animals. First, we discuss the clinical classification and pathomechanism of TBI, the research in animal models, and the neuroprotective role of E2. Based on the results of animal studies and clinical trials, we discuss possible E2 targets from early to late events in the pathomechanism of TBI, including neuroinflammation and possible disturbances of the endocrine system. Finally, the potential relevance of selective estrogenic compounds in the treatment of TBI will be discussed.
Collapse
Affiliation(s)
- Erzsébet Kövesdi
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Center for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pecs, Hungary;
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pecs, Hungary;
| | - István M. Abrahám
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Center for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pecs, Hungary;
| |
Collapse
|
64
|
An update on the association between traumatic brain injury and Alzheimer's disease: Focus on Tau pathology and synaptic dysfunction. Neurosci Biobehav Rev 2020; 120:372-386. [PMID: 33171143 DOI: 10.1016/j.neubiorev.2020.10.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
L.P. Li, J.W. Liang and H.J. Fu. An update on the association between traumatic brain injury and Alzheimer's disease: Focus on Tau pathology and synaptic dysfunction. NEUROSCI BIOBEHAV REVXXX-XXX,2020.-Traumatic brain injury (TBI) and Alzheimer's disease (AD) are devastating conditions that have long-term consequences on individual's cognitive functions. Although TBI has been considered a risk factor for the development of AD, the link between TBI and AD is still in debate. Aggregation of hyperphosphorylated tau and intercorrelated synaptic dysfunction, two key pathological elements in both TBI and AD, play a pivotal role in mediating neurodegeneration and cognitive deficits, providing a mechanistic link between these two diseases. In the first part of this review, we analyze the experimental literatures on tau pathology in various TBI models and review the distribution, biological features and mechanisms of tau pathology following TBI with implications in AD pathogenesis. In the second part, we review evidences of TBI-mediated structural and functional impairments in synapses, with a focus on the overlapped mechanisms underlying synaptic abnormalities in both TBI and AD. Finally, future perspectives are proposed for uncovering the complex relationship between TBI and neurodegeneration, and developing potential therapeutic avenues for alleviating cognitive deficits after TBI.
Collapse
|
65
|
Acute damage to the blood–brain barrier and perineuronal net integrity in a clinically-relevant rat model of traumatic brain injury. Neuroreport 2020; 31:1167-1174. [PMID: 32991524 DOI: 10.1097/wnr.0000000000001531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Closed-head, frontal impacts in which the brain undergoes both lateral and rotational acceleration comprise the majority of human traumatic brain injury (TBI). Here, we utilize a clinically relevant model to examine the effects of a single concussion on aspects of brain integrity: the blood-brain barrier, the perineuronal nets (PNNs), and diffuse axonal injury. Adult, male Sprague-Dawley rats received either a frontal, closed-head concussive TBI, or no injury and were evaluated at 1- or 7-day post-injury. Using immunolabeling for albumin, we observed a significant increase in the permeability of the blood-brain barrier at 1-, but not 7-day post-injury. Breakdown of the PNN, as measured by the binding of wisteria floribunda, was observed at 1-day post-injury in the dorsal, lateral, and ventral cortices. This difference was resolved at 7-day. Finally, axonal injury was identified at both 1- and 7-day post-injury. This preclinical model of closed-head, frontal TBI presents a useful tool with which to understand better the acute pathophysiology of a single, frontal TBI.
Collapse
|
66
|
Li H, Lu C, Yao W, Xu L, Zhou J, Zheng B. Dexmedetomidine inhibits inflammatory response and autophagy through the circLrp1b/miR-27a-3p/Dram2 pathway in a rat model of traumatic brain injury. Aging (Albany NY) 2020; 12:21687-21705. [PMID: 33147167 PMCID: PMC7695368 DOI: 10.18632/aging.103975] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022]
Abstract
Circular RNAs (circRNAs) have a regulatory function on inflammation and autophagy, of which rno-circRNA_010705 (circLrp1b) appears to be significantly up-regulated following traumatic brain injury (TBI). Dexmedetomidine (DEX) shows improvement effects in TBI by inhibiting NLRP3/caspase-1. However, whether circLrp1b plays critical roles in DEX-mediated TBI attenuation and the underlying mechanisms remain unclear. After TBI was established in rats by controlled cortical impact (CCI) to cause brain trauma, they received an intracerebroventricular injection of lentiviral vector, followed by intraperitoneal injection of DEX. Administration of DEX ameliorated autophagy in rats following TBI, accompanied by up-regulated circLrp1b and Dram2 and down-regulated miR-27a-3p. DEX promoted the effects of circLrp1b in attenuating TBI-induced neurologic impairment, autophagy, and inflammation, which was significantly reversed by inhibition of miR-27a-3p or Dram2 overexpression. Mechanistically, northern blot and luciferase reporter assays indicated that circLrp1b up-regulated Dram2 expression by functioning as a sponge for miR-27a-3p to promote autophagy involved in TBI, which was reversed by DEX treatment. Collectively, this study demonstrated that DEX inhibits inflammatory response and autophagy involved in TBI in vivo through inactivation of the circLrp1b/miR-27a-3p/Dram2 signaling pathway.
Collapse
Affiliation(s)
- Hengchang Li
- Department of Anesthesiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Chengxiang Lu
- Department of Anesthesiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Wenfei Yao
- Department of Anesthesiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Lixin Xu
- Department of Anesthesiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jun Zhou
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Bin Zheng
- Department of Anesthesiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
67
|
Begemann MJ, Brand BA, Ćurčić-Blake B, Aleman A, Sommer IE. Efficacy of non-invasive brain stimulation on cognitive functioning in brain disorders: a meta-analysis. Psychol Med 2020; 50:2465-2486. [PMID: 33070785 PMCID: PMC7737055 DOI: 10.1017/s0033291720003670] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/27/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cognition is commonly affected in brain disorders. Non-invasive brain stimulation (NIBS) may have procognitive effects, with high tolerability. This meta-analysis evaluates the efficacy of transcranial magnetic stimulation (TMS) and transcranial Direct Current Stimulation (tDCS) in improving cognition, in schizophrenia, depression, dementia, Parkinson's disease, stroke, traumatic brain injury, and multiple sclerosis. METHODS A PRISMA systematic search was conducted for randomized controlled trials. Hedges' g was used to quantify effect sizes (ES) for changes in cognition after TMS/tDCS v. sham. As different cognitive functions may have unequal susceptibility to TMS/tDCS, we separately evaluated the effects on: attention/vigilance, working memory, executive functioning, processing speed, verbal fluency, verbal learning, and social cognition. RESULTS We included 82 studies (n = 2784). For working memory, both TMS (ES = 0.17, p = 0.015) and tDCS (ES = 0.17, p = 0.021) showed small but significant effects. Age positively moderated the effect of TMS. TDCS was superior to sham for attention/vigilance (ES = 0.20, p = 0.020). These significant effects did not differ across the type of brain disorder. Results were not significant for the other five cognitive domains. CONCLUSIONS Our results revealed that both TMS and tDCS elicit a small trans-diagnostic effect on working memory, tDCS also improved attention/vigilance across diagnoses. Effects on the other domains were not significant. Observed ES were small, yet even slight cognitive improvements may facilitate daily functioning. While NIBS can be a well-tolerated treatment, its effects appear domain specific and should be applied only for realistic indications (i.e. to induce a small improvement in working memory or attention).
Collapse
Affiliation(s)
- Marieke J. Begemann
- Department of Biomedical Sciences of Cells & Systems, Section Cognitive Neurosciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bodyl A. Brand
- Department of Biomedical Sciences of Cells & Systems, Section Cognitive Neurosciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Branislava Ćurčić-Blake
- Department of Biomedical Sciences of Cells & Systems, Section Cognitive Neurosciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - André Aleman
- Department of Biomedical Sciences of Cells & Systems, Section Cognitive Neurosciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Iris E. Sommer
- Department of Biomedical Sciences of Cells & Systems, Section Cognitive Neurosciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
68
|
Liu F, Huang J, Hei G, Wu R, Liu Z. Effects of sulforaphane on cognitive function in patients with frontal brain damage: study protocol for a randomised controlled trial. BMJ Open 2020; 10:e037543. [PMID: 33067279 PMCID: PMC7569949 DOI: 10.1136/bmjopen-2020-037543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Many patients with frontal brain damage show serious cognitive function deficits, which hamper their quality of life and result in poor clinical outcomes. Preclinical research has shown that sulforaphane can significantly improve spatial localisation and working memory impairment after brain injury. The primary aim of this double-blind randomised controlled clinical trial is to assess the efficacy of sulforaphane for improving cognitive function in patients with frontal brain damage. METHODS AND ANALYSIS Ninety eligible patients will be randomly allocated to an active treatment or a placebo group in a 2:1 ratio. Participants will undergo a series of cognitive and neuropsychiatric tests at baseline (week 0) and after 12 weeks to determine the effect of sulforaphane on cognition. Magnetic resonance spectrum of the brain will be studied using the 3T MRIs of the brain to detect brain metabolites markers, including N-acetyl aspartate, glutamate (Glu), glutathione (GSH) and γ-aminobutyric acid (GABA). Blood brain-derived neurotrophic factor, Glu, GSH and GABA levels and gut microbiota will also be assessed over this period. This study will also evaluate long-term outcomes of brain trauma, brain tumours and cerebrovascular disease via exploratory analyses. The primary outcome will be the difference in scores of a battery of cognitive tests after 12 weeks of sulforaphane treatment. The secondary outcomes will be changes in the Functional Activities Questionnaire (FAQ), the Patient Health Questionnaire (PHQ-9), the Self-Rating Anxiety Scale, the changes in T1-weighted MRI and resting-state functional MRI findings, and changes in brain and blood metabolic markers and gut microbiota at weeks 0 and 12. We expect that sulforaphane will yield favourable results in treating memory and learning deficits for patients with frontal brain damage. Cognitive functional treatment may also improve brain trauma, brain tumours and cerebrovascular outcomes. ETHICS AND DISSEMINATION The study protocol has been approved by the Medical Ethics committee of the Xiangya Hospital of Central South University (No. 2017121019). The results will be disseminated in peer-reviewed journals and at international conferences. TRIAL REGISTRATION NUMBER This trial was registered on Clinicaltrials.gov on 31 January 2020 (NCT04252261). The protocol version is V.1.0 (20 December 2019).
Collapse
Affiliation(s)
- Fangkun Liu
- Department of Neurosurgery, Central South University (CSU), 410008, Changsha, Hunan, China
| | - Jing Huang
- Department of Psychiatry, the Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China; Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center on Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, 410011, Changsha, Hunan, China
| | - Gangrui Hei
- Department of Psychiatry, the Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China; Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center on Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, 410011, Changsha, Hunan, China
| | - Renrong Wu
- Department of Psychiatry, the Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China; Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center on Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, 410011, Changsha, Hunan, China
| | - Zhixiong Liu
- Department of Neurosurgery, Central South University (CSU), 410008, Changsha, Hunan, China
| |
Collapse
|
69
|
Liu Y, Guo C, Ding Y, Long X, Li W, Ke D, Wang Q, Liu R, Wang JZ, Zhang H, Wang X. Blockage of AEP attenuates TBI-induced tau hyperphosphorylation and cognitive impairments in rats. Aging (Albany NY) 2020; 12:19421-19439. [PMID: 33040048 PMCID: PMC7732271 DOI: 10.18632/aging.103841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/29/2020] [Indexed: 01/24/2023]
Abstract
Traumatic brain injury (TBI) is regarded as a high-risk factor for Alzheimer's disease (AD). Asparaginyl endopeptidase (AEP), a lysosomal cysteine protease involved in AD pathogenesis, is normally activated under acidic conditions and also in TBI. However, both the molecular mechanism underlying AEP activation-mediated TBI-related AD pathologies, and the role of AEP as an AD therapeutic target, still remain unclear. Here, we report that TBI induces hippocampus dependent cognitive deficit and synaptic dysfunction, accompanied with AEP activation, I2PP2A (inhibitor 2 of PP2A, also called SET) mis-translocation from neuronal nucleus to cytoplasm, an obvious increase in AEP interaction with SET, and tau hyperphosphorylation in hippocampus of rats. Oxygen-glucose deprivation (OGD), mimicking an acidic condition, also leads to AEP activation, SET mis-translocation, PP2A inhibition, tau hyperphosphorylation, and a decrease in synaptic proteins, all of which are abrogated by AEP inhibitor AENK in primary neurons. Interestingly, AENK restores SET back to the nucleus, mitigates tau pathologies, rescuing TBI-induced cognitive deficit in rats. These findings highlight a novel etiopathogenic mechanism of TBI-related AD, which is initiated by AEP activation, accumulating SET in cytoplasm, and favoring tau pathology and cognitive impairments. Lowering AEP activity by AEP inhibitor would be beneficial to AD patients with TBI.
Collapse
Affiliation(s)
- Yi Liu
- Department of Pathophysiology, Weifang Medical University, Weifang 261053, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cuiping Guo
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Ding
- Department of Pathophysiology, Weifang Medical University, Weifang 261053, China
| | - Xiaobing Long
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, China
| | - Wensheng Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, JS, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, JS, China
| |
Collapse
|
70
|
Abstract
Elevated cholesterol is a major risk factor in the development of cardiovascular disease. Statins have proven to be effective in lowering low-density lipoprotein cholesterol as well as the incidence of cardiovascular events. As a result, statins are widely prescribed in the United States, with an estimated 35 million patients on statins. Many of these patients are older than age 65 and suffer from various comorbidities, including mild to severe cognitive impairment. Early studies looking at the effects of statins on cognition have shown that statin use may lead to mild reversible cognitive decline, although long-term studies have shown inconclusive findings. In recent years, studies have shown that the use of statins in certain groups of patients may lead to a reduction in the rate of cognitive decline. One hypothesis for this finding is that statin use can reduce the risk of cerebrovascular disease which may, in turn, reduce the risk of mild cognitive decline and dementia. With numerous patients currently prescribed statins and the likelihood that more patients will be prescribed the medication in the coming years, it is important to review the current literature to determine the association between statin use and cognitive decline, as well as determine how statins may be beneficial in preventing cognitive decline.
Collapse
Affiliation(s)
- Bhawneet Chadha
- From the Department of Medicine, Tufts Medical Center, Boston, MA
| | - William H Frishman
- Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| |
Collapse
|
71
|
Neves K, Guercio GD, Anjos-Travassos Y, Costa S, Perozzo A, Montezuma K, Herculano-Houzel S, Panizzutti R. The relationship between the number of neurons and behavioral performance in Swiss mice. Neurosci Lett 2020; 735:135202. [PMID: 32599318 DOI: 10.1016/j.neulet.2020.135202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/21/2020] [Indexed: 10/24/2022]
Abstract
Neuronal number varies by several orders of magnitude across species, and has been proposed to predict cognitive capability across species. Remarkably, numbers of neurons vary across individual mice by a factor of 2 or more. We directly addressed the question of whether there is a relationship between performance in behavioral tests and the number of neurons in functionally relevant structures in the mouse brain. Naïve Swiss mice went through a battery of behavioral tasks designed to measure cognitive, motor and olfactory skills. We estimated the number of neurons in different brain regions (cerebral cortex, hippocampus, olfactory bulb, cerebellum and remaining areas) and crossed the two datasets to test the a priori hypothesis of correlation between cognitive abilities and numbers of neurons. Surprisingly, performance in the behavioral tasks did not correlate strongly with number of neurons in any of the brain regions studied. Our results show that whereas neuronal number is a predictor of cognitive skills across species, it is not a good predictor of cognitive, sensory or motor ability across individuals within a species, which suggests that other factors are more relevant for explaining cognitive differences between individuals of the same species.
Collapse
Affiliation(s)
- Kleber Neves
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brazil; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil.
| | - Gerson Duarte Guercio
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Department of Psychiatry, University of Minnesota, 2312 S 6th St., Minneapolis, MN, United States.
| | - Yuri Anjos-Travassos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Stella Costa
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Ananda Perozzo
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Karine Montezuma
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Suzana Herculano-Houzel
- Department of Psychology, Department of Biological Sciences, Vanderbilt Brain Institute, Vanderbilt University, United States
| | - Rogério Panizzutti
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Instituto de Psiquiatria, Universidade Federal do Rio de Janeiro, Brazil
| |
Collapse
|
72
|
Xin D, Li T, Chu X, Ke H, Yu Z, Cao L, Bai X, Liu D, Wang Z. Mesenchymal stromal cell-derived extracellular vesicles modulate microglia/macrophage polarization and protect the brain against hypoxia-ischemic injury in neonatal mice by targeting delivery of miR-21a-5p. Acta Biomater 2020; 113:597-613. [PMID: 32619670 DOI: 10.1016/j.actbio.2020.06.037] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/01/2020] [Accepted: 06/25/2020] [Indexed: 12/19/2022]
Abstract
Mesenchymal stromal cell (MSC)-derived extracellular vesicles (EVs) (MSC-EVs) exhibit protective effects in damaged or diseased tissues. However, the role of EVs secreted by MSC in hypoxia-ischemic (HI) injury in neonatal mice remains unknown. Systemic administration of MSC-EVs attenuated acute brain damage and neuroinflammation, and skewed CD11b+/CD45low microglia and CD11b+/CD45high brain monocyte/macrophage towards a more anti-inflammatory property as determined at 72 h post-HI. In addition, MSC-EVs remarkably improve the injury outcomes pups prior to weaning (P21), while no effect on long-term memory impairment (P42). Importantly, these effects were preceded by incorporation of MSC-EVs into a large number of neurons and microglia within HI group. Abundant levels of miR-21a-5p were present in EVs as determined with next-generation sequencing. Notably, MSC-EVs treatment further increased miR-21a-5p levels at 72 h post HI. Knockdown analyses revealed that miR-21a-5p, and its target-Timp3, were essential for this neuroprotective property of MSC-EVs following HI exposure as demonstrated in both in vitro and in vivo models. These findings suggest that a systemic administration of EVs derived from MSC, have the capacity to incorporated into neurons and microglia where they can then exert neuroprotection against HI-induced injury in neonates through the delivery of miR-21a-5p.
Collapse
Affiliation(s)
- Danqing Xin
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Tingting Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Xili Chu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Hongfei Ke
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Zhuoya Yu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Lili Cao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Xuemei Bai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
73
|
Sauerbeck AD, Gangolli M, Reitz SJ, Salyards MH, Kim SH, Hemingway C, Gratuze M, Makkapati T, Kerschensteiner M, Holtzman DM, Brody DL, Kummer TT. SEQUIN Multiscale Imaging of Mammalian Central Synapses Reveals Loss of Synaptic Connectivity Resulting from Diffuse Traumatic Brain Injury. Neuron 2020; 107:257-273.e5. [PMID: 32392471 PMCID: PMC7381374 DOI: 10.1016/j.neuron.2020.04.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/04/2020] [Accepted: 04/11/2020] [Indexed: 02/07/2023]
Abstract
The brain's complex microconnectivity underlies its computational abilities and vulnerability to injury and disease. It has been challenging to illuminate the features of this synaptic network due to the small size and dense packing of its elements. Here, we describe a rapid, accessible super-resolution imaging and analysis workflow-SEQUIN-that quantifies central synapses in human tissue and animal models, characterizes their nanostructural and molecular features, and enables volumetric imaging of mesoscale synaptic networks without the production of large histological arrays. Using SEQUIN, we identify cortical synapse loss resulting from diffuse traumatic brain injury, a highly prevalent connectional disorder. Similar synapse loss is observed in three murine models of Alzheimer-related neurodegeneration, where SEQUIN mesoscale mapping identifies regional synaptic vulnerability. These results establish an easily implemented and robust nano-to-mesoscale synapse quantification and characterization method. They furthermore identify a shared mechanism-synaptopathy-between Alzheimer neurodegeneration and its best-established epigenetic risk factor, brain trauma.
Collapse
Affiliation(s)
- Andrew D Sauerbeck
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mihika Gangolli
- McKelvey School of Engineering, Washington University, St. Louis, MO 63130, USA; Currently, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sydney J Reitz
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maverick H Salyards
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel H Kim
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher Hemingway
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians Universität München, Munich 82152, Germany
| | - Maud Gratuze
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tejaswi Makkapati
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians Universität München, Munich 82152, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich 81377, Germany
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David L Brody
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Currently, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Terrance T Kummer
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
74
|
Saikumar J, Byrns CN, Hemphill M, Meaney DF, Bonini NM. Dynamic neural and glial responses of a head-specific model for traumatic brain injury in Drosophila. Proc Natl Acad Sci U S A 2020; 117:17269-17277. [PMID: 32611818 PMCID: PMC7382229 DOI: 10.1073/pnas.2003909117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is the strongest environmental risk factor for the accelerated development of neurodegenerative diseases. There are currently no therapeutics to address this due to lack of insight into mechanisms of injury progression, which are challenging to study in mammalian models. Here, we have developed and extensively characterized a head-specific approach to TBI in Drosophila, a powerful genetic system that shares many conserved genes and pathways with humans. The Drosophila TBI (dTBI) device inflicts mild, moderate, or severe brain trauma by precise compression of the head using a piezoelectric actuator. Head-injured animals display features characteristic of mammalian TBI, including severity-dependent ataxia, life span reduction, and brain degeneration. Severe dTBI is associated with cognitive decline and transient glial dysfunction, and stimulates antioxidant, proteasome, and chaperone activity. Moreover, genetic or environmental augmentation of the stress response protects from severe dTBI-induced brain degeneration and life span deficits. Together, these findings present a tunable, head-specific approach for TBI in Drosophila that recapitulates mammalian injury phenotypes and underscores the ability of the stress response to mitigate TBI-induced brain degeneration.
Collapse
Affiliation(s)
- Janani Saikumar
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - China N Byrns
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Matthew Hemphill
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104;
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
75
|
Mechanisms of cell damage due to mechanical impact: an in vitro investigation. Sci Rep 2020; 10:12009. [PMID: 32686715 PMCID: PMC7371734 DOI: 10.1038/s41598-020-68655-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/30/2020] [Indexed: 01/07/2023] Open
Abstract
The dynamic response of cells when subjected to mechanical impact has become increasingly relevant for accurate assessment of potential blunt injuries and elucidating underlying injury mechanisms. When exposed to mechanical impact, a biological system such as the human skin, brain, or liver is rapidly accelerated, which could result in blunt injuries. For this reason, an acceleration of greater than > 150 g is the most commonly used criteria for head injury. To understand the main mechanism(s) of blunt injury under such extreme dynamic threats, we have developed an innovative experimental method that applies a well-characterized and -controlled mechanical impact to live cells cultured in a custom-built in vitro setup compatible with live cell microscopy. Our studies using fibroblast cells as a model indicate that input acceleration (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${a}_{in}$$\end{document}ain) alone, even when it is much greater than the typical injury criteria, e.g., \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${a}_{in}>1{,}000$$\end{document}ain>1,000 g, does not result in cell damage. On the contrary, we have observed a material-dependent critical pressure value above which a sudden decrease in cell population and cell membrane damage have been observed. We have unambiguously shown that (1) this critical pressure is associated with the onset of cavitation bubbles in a cell culture chamber and (2) the dynamics of cavitation bubbles in the chamber induces localized compressive/tensile pressure cycles, with an amplitude that is considerably greater than the acceleration-induced pressure, to cells. More importantly, the rate of pressure change with time for cavitation-induced pressure is significantly faster (more than ten times) than acceleration-induced pressure. Our in vitro study on the dynamic response of biological systems due to mechanical impact is a crucial step towards understanding potential mechanism(s) of blunt injury and implementing novel therapeutic strategies post-trauma.
Collapse
|
76
|
Transport rate of EAAT2 is regulated by amino acid located at the interface between the scaffolding and substrate transport domains. Neurochem Int 2020; 139:104792. [PMID: 32668264 DOI: 10.1016/j.neuint.2020.104792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/19/2020] [Accepted: 06/21/2020] [Indexed: 01/24/2023]
Abstract
Excitatory Amino Acid Transporters (EAATs) are plasma membrane proteins responsible for maintenance of low extracellular concentrations of glutamate in the CNS. Dysfunction in their activity is implicated in various neurological disorders. Glutamate transport by EAATs occurs through the movement of the central transport domain relative to the scaffold domain in the EAAT membrane protein. Previous studies suggested that residues located within the interface of these two domains in EAAT2, the main subtype of glutamate transporter in the brain, are involved in regulating transport rates. We used mutagenesis, structure-function relationship, surface protein expression and electrophysiology studies, in transfected COS-7 cells and oocytes, to examine residue glycine at position 298, which is located within this interface. Mutation G298A results in increased transport rate without changes in surface expression, suggesting a more hydrophobic and larger alanine results in facilitated transport movement. The increased transport rate does not involve changes in sodium affinity. Electrophysiological currents show that G298A increase both transport and anion currents, suggesting faster transitions through the transport cycle. This work identifies a region critically involved in setting the glutamate transport rate.
Collapse
|
77
|
Liu B, Cao Y, Shi F, Wang L, Li N, Cheng X, Du J, Tian Q, Zhou X. The overexpression of RBM3 alleviates TBI-induced behaviour impairment and AD-like tauopathy in mice. J Cell Mol Med 2020; 24:9176-9188. [PMID: 32648620 PMCID: PMC7417709 DOI: 10.1111/jcmm.15555] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/08/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
The therapeutic hypothermia is an effective tool for TBI‐associated brain impairment, but its side effects limit in clinical routine use. Hypothermia up‐regulates RNA‐binding motif protein 3 (RBM3), which is verified to protect synaptic plasticity. Here, we found that cognitive and LTP deficits, loss of spines, AD‐like tau pathologies are displayed one month after TBI in mice. In contrast, the deficits of LTP and cognitive, loss of spines and tau abnormal phosphorylation at several sites are obviously reversed in TBI mice combined with hypothermia pre‐treatment (HT). But, the neuroprotective role of HT disappears in TBI mouse models under condition of blocking RBM3 expression with RBM3 shRNA. In other hand, overexpressing RBM3 by AAV‐RBM3 plasmid can mimic HT‐like neuroprotection against TBI‐induced chronic brain injuries, such as improving LTP and cognitive, loss of spines and tau hyperphosphorylation in TBI mouse models. Taken together, hypothermia pre‐treatment reverses TBI‐induced chronic AD‐like pathology and behaviour deficits in RBM3 expression dependent manner, RBM3 may be a potential target for neurodegeneration diseases including Alzheimer disease.
Collapse
Affiliation(s)
- Bingjin Liu
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Medicine and Pharmaceutical Engineering, Taizhou Vocational and Technical College, Taizhou, China
| | - Yun Cao
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangxiao Shi
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Wang
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Li
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangshu Cheng
- Department of Neurology, Center for Translational Medicine, Huaihe Hospital of Henan University, Kaifeng, China
| | - Jin Du
- Department of Neurology, Center for Translational Medicine, Huaihe Hospital of Henan University, Kaifeng, China
| | - Qing Tian
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinwen Zhou
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
78
|
Selvakumar GP, Ahmed ME, Iyer SS, Thangavel R, Kempuraj D, Raikwar SP, Bazley K, Wu K, Khan A, Kukulka K, Bussinger B, Zaheer S, Burton C, James D, Zaheer A. Absence of Glia Maturation Factor Protects from Axonal Injury and Motor Behavioral Impairments after Traumatic Brain Injury. Exp Neurobiol 2020; 29:230-248. [PMID: 32565489 PMCID: PMC7344375 DOI: 10.5607/en20017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) causes disability and death, accelerating the progression towards Alzheimer's disease and Parkinson's disease (PD). TBI causes serious motor and cognitive impairments, as seen in PD that arise during the period of the initial insult. However, this has been understudied relative to TBI induced neuroinflammation, motor and cognitive decline that progress towards PD. Neuronal ubiquitin-C-terminal hydrolase- L1 (UCHL1) is a thiol protease that breaks down ubiquitinated proteins and its level represents the severity of TBI. Previously, we demonstrated the molecular action of glia maturation factor (GMF); a proinflammatory protein in mediating neuroinflammation and neuronal loss. Here, we show that the weight drop method induced TBI neuropathology using behavioral tests, western blotting, and immunofluorescence techniques on sections from wild type (WT) and GMF-deficient (GMF-KO) mice. Results reveal a significant improvement in substantia nigral tyrosine hydroxylase and dopamine transporter expression with motor behavioral performance in GMF-KO mice following TBI. In addition, a significant reduction in neuroinflammation was manifested, as shown by activation of nuclear factor-kB, reduced levels of inducible nitric oxide synthase, and cyclooxygenase- 2 expressions. Likewise, neurotrophins including brain-derived neurotrophic factor and glial-derived neurotrophic factor were significantly improved in GMF-KO mice than WT 72 h post-TBI. Consistently, we found that TBI enhances GFAP and UCHL-1 expression and reduces the number of dopaminergic TH-positive neurons in WT compared to GMF-KO mice 72 h post-TBI. Interestingly, we observed a reduction of THpositive tanycytes in the median eminence of WT than GMF-KO mice. Overall, we found that absence of GMF significantly reversed these neuropathological events and improved behavioral outcome. This study provides evidence that PD-associated pathology progression can be initiated upon induction of TBI.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Klaudia Kukulka
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Bret Bussinger
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
79
|
In Vivo Diffusion Tensor Imaging in Acute and Subacute Phases of Mild Traumatic Brain Injury in Rats. eNeuro 2020; 7:ENEURO.0476-19.2020. [PMID: 32424056 PMCID: PMC7307627 DOI: 10.1523/eneuro.0476-19.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 04/27/2020] [Accepted: 05/11/2020] [Indexed: 12/23/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is the most common form of TBI with 10–25% of the patients experiencing long-lasting symptoms. The potential of diffusion tensor imaging (DTI) for evaluating microstructural damage after TBI is widely recognized, but the interpretation of DTI changes and their relationship with the underlying tissue damage is unclear. We studied how both axonal damage and gliosis contribute to DTI alterations after mTBI. We induced mTBI using the lateral fluid percussion (LFP) injury model in adult male Sprague Dawley rats and scanned them at 3 and 28 d post-mTBI. To characterize the DTI findings in the tissue, we assessed the histology by performing structure tensor (ST)-based analysis and cell counting on myelin-stained and Nissl-stained sections, respectively. In particular, we studied the contribution of two tissue components, myelinated axons and cellularity, to the DTI changes. Fractional anisotropy (FA), mean diffusivity (MD), and axial diffusivity (AD) were decreased in both white and gray matter areas in the acute phase post-mTBI, mainly at the primary lesion site. In the subacute phase, FA and AD were decreased in the white matter, external capsule, corpus callosum, and internal capsule. Our quantitative histologic assessment revealed axonal damage and gliosis throughout the brain in both white and gray matter, consistent with the FA and AD changes. Our findings suggest that the usefulness of in vivo DTI is limited in its detection of secondary damage distal to the primary lesion, while at the lesion site, DTI detected progressive microstructural damage in the white and gray matter after mTBI.
Collapse
|
80
|
Nguyen LD, Ehrlich BE. Cellular mechanisms and treatments for chemobrain: insight from aging and neurodegenerative diseases. EMBO Mol Med 2020; 12:e12075. [PMID: 32346964 PMCID: PMC7278555 DOI: 10.15252/emmm.202012075] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/09/2020] [Accepted: 04/01/2020] [Indexed: 12/22/2022] Open
Abstract
Chemotherapy is a life-saving treatment for cancer patients, but also causes long-term cognitive impairment, or "chemobrain", in survivors. However, several challenges, including imprecise diagnosis criteria, multiple confounding factors, and unclear and heterogeneous molecular mechanisms, impede effective investigation of preventions and treatments for chemobrain. With the rapid increase in the number of cancer survivors, chemobrain is an urgent but unmet clinical need. Here, we leverage the extensive knowledge in various fields of neuroscience to gain insights into the mechanisms for chemobrain. We start by outlining why the post-mitotic adult brain is particularly vulnerable to chemotherapy. Next, through drawing comparisons with normal aging, Alzheimer's disease, and traumatic brain injury, we identify universal cellular mechanisms that may underlie the cognitive deficits in chemobrain. We further identify existing neurological drugs targeting these cellular mechanisms that can be repurposed as treatments for chemobrain, some of which were already shown to be effective in animal models. Finally, we briefly describe future steps to further advance our understanding of chemobrain and facilitate the development of effective preventions and treatments.
Collapse
Affiliation(s)
- Lien D Nguyen
- Department of Pharmacology and Interdepartmental Neuroscience ProgramYale UniversityNew HavenCTUSA
| | - Barbara E Ehrlich
- Department of Pharmacology and Interdepartmental Neuroscience ProgramYale UniversityNew HavenCTUSA
| |
Collapse
|
81
|
Mira RG, Lira M, Quintanilla RA, Cerpa W. Alcohol consumption during adolescence alters the hippocampal response to traumatic brain injury. Biochem Biophys Res Commun 2020; 528:514-519. [PMID: 32505350 DOI: 10.1016/j.bbrc.2020.05.160] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/21/2020] [Indexed: 01/08/2023]
Abstract
Binge drinking is the consumption of large volumes of alcohol in short periods and exerts its effects on the central nervous system, including the hippocampus. We have previously shown that binge drinking alters mitochondrial dynamics and induces neuroinflammation in the hippocampus of adolescent rats. Mild traumatic brain injury (mTBI), is regularly linked to alcohol consumption and share mechanisms of brain damage. In this context, we hypothesized that adolescent binge drinking could prime the development of brain damage generated by mTBI. We found that alcohol binge drinking induced by the "drinking in the dark" (DID) paradigm increases oxidative damage and astrocyte activation in the hippocampus of adolescent mice. Interestingly, adolescent animals submitted to DID showed decreased levels of mitofusin 2 that controls mitochondrial dynamics. When mTBI was evaluated as a second challenge, hippocampi from animals previously submitted to DID showed a reduction in dendritic spine number and a different spine profile. Mitochondrial performance could be compromised by alterations in mitochondrial fission in DID-mTBI animals. These data suggest that adolescent alcohol consumption can modify the progression of mTBI pathophysiology. We propose that mitochondrial impairment and oxidative damage could act as priming factors, modifying predisposition against mTBI effects.
Collapse
Affiliation(s)
- Rodrigo G Mira
- Laboratorio de función y patología neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile
| | - Matías Lira
- Laboratorio de función y patología neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo A Quintanilla
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile; Laboratorio de Enfermedades Neurodegenerativas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Waldo Cerpa
- Laboratorio de función y patología neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
82
|
Rashno M, Ghaderi S, Nesari A, Khorsandi L, Farbood Y, Sarkaki A. Chrysin attenuates traumatic brain injury-induced recognition memory decline, and anxiety/depression-like behaviors in rats: Insights into underlying mechanisms. Psychopharmacology (Berl) 2020; 237:1607-1619. [PMID: 32088834 DOI: 10.1007/s00213-020-05482-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 02/12/2020] [Indexed: 12/20/2022]
Abstract
RATIONALE Cortical and hippocampal neuronal apoptosis and neuroinflammation are associated with behavioral deficits following traumatic brain injury (TBI). OBJECTIVES The present study was designed to investigate the potential protective effects of flavonoid chrysin against TBI-induced vestibulomotor impairment, exploratory/locomotor dysfunctions, recognition memory decline, and anxiety/depression-like behaviors, as well as the verified possible involved mechanisms. METHODS Chrysin (25, 50, or 100 mg/kg/day; P.O.) was administered to rats immediately after diffuse TBI induction, and it was continued for 3 or 14 days. Behavioral functions were assessed by employing standard behavioral paradigms at scheduled points in time. Three days post-TBI, inflammation status was assayed in both cerebral cortex and hippocampus using ELISA kits. Moreover, apoptosis and expression of Bcl-2 family proteins were examined by TUNEL staining and immunohistochemistry, respectively. RESULTS The results indicated that treatment with chrysin improved vestibulomotor dysfunction, ameliorated recognition memory deficit, and attenuated anxiety/depression-like behaviors in the rats with TBI. Chrysin treatment also modulated inflammation status, reduced apoptotic index, and regulated Bcl-2 family proteins expression in the brains of rats with TBI. CONCLUSIONS In conclusion, the results suggest that chrysin could be beneficial for protection against TBI-associated behavioral deficits, owing to its anti-apoptotic and anti-inflammatory properties.
Collapse
Affiliation(s)
- Masome Rashno
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahab Ghaderi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Nesari
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoob Farbood
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Sarkaki
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
83
|
Mohammadi M, Manaheji H, Maghsoudi N, Danyali S, Baniasadi M, Zaringhalam J. Microglia dependent BDNF and proBDNF can impair spatial memory performance during persistent inflammatory pain. Behav Brain Res 2020; 390:112683. [PMID: 32442548 DOI: 10.1016/j.bbr.2020.112683] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/15/2020] [Accepted: 04/27/2020] [Indexed: 12/30/2022]
Abstract
Inflammatory pain is commonly associated with cognitive impairment. However, its molecular mechanisms are poorly understood. Thus, this study was conducted to investigate the molecular mechanisms of behavioral changes associated with inflammatory pain. Briefly, 36 Wistar rats were randomly divided into two main groups: CFA group treated with 100 μL of Complete Freunds' Adjuvant (CFA) and CFA + Minocycline group treated with 100 μL of CFA+40 mg/kg/day of minocycline). After that, each group was divided into three subgroups based on different time points of the study. The pain was induced using CFA and subsequent behavioral changes (i.e., hyperalgesia and learning and spatial memory) were analyzed by the Morris Water Maze (MWM) task and Radiant Heat. Then, the cellular and molecular changes were assessed using Western Blotting, Immunohistochemistry, and Terminal deoxynucleotidyl transferase dUTP Nick End Labeling (TUNEL) techniques. Results of the study indicated that CFA-induced pain impaired spatial learning and memory functions. Studying the cellular changes showed that persistent inflammatory pain increased the microglial activity in CA1 and Dentate Gyrus (DG) regions. Furthermore, an increase was observed in the percentage of TUNEL-positive cells. Also, pro-Brain-Derived Neurotrophic Factor (BDNF)/BDNF ratio, Caspase3, and Receptor-Interacting Protein kinase 3 (RIP3) levels increased in the rats' hippocampus following induction of persistent inflammatory pain. These changes were reversed following the cessation of pain as well as the injection of minocycline. Taking together, the results of the current study for the first time revealed that an increase in the microglia dependent proBDNF/BDNF ratio following persistent inflammatory pain leads to cell death of the CA1 and DG neurons that subsequently causes a cognitive deficit in the learning and spatial memory functions.
Collapse
Affiliation(s)
- Mola Mohammadi
- Physiology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homa Manaheji
- Physiology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Maghsoudi
- Department of Biology, Queens College and Graduate Center of the City University of New York, Flushing, NY, USA; Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Danyali
- Physiology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mansoureh Baniasadi
- Physiology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalal Zaringhalam
- Physiology Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
84
|
Marquez AM, Morgan RW, Ko T, Landis WP, Hefti MM, Mavroudis CD, McManus MJ, Karlsson M, Starr J, Roberts AL, Lin Y, Nadkarni V, Licht DJ, Berg RA, Sutton RM, Kilbaugh TJ. Oxygen Exposure During Cardiopulmonary Resuscitation Is Associated With Cerebral Oxidative Injury in a Randomized, Blinded, Controlled, Preclinical Trial. J Am Heart Assoc 2020; 9:e015032. [PMID: 32321350 PMCID: PMC7428577 DOI: 10.1161/jaha.119.015032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Hyperoxia during cardiopulmonary resuscitation (CPR) may lead to oxidative injury from mitochondrial‐derived reactive oxygen species, despite guidelines recommending 1.0 inspired oxygen during CPR. We hypothesized exposure to 1.0 inspired oxygen during CPR would result in cerebral hyperoxia, higher mitochondrial‐derived reactive oxygen species, increased oxidative injury, and similar survival compared with those exposed to 21% oxygen. Methods and Results Four‐week‐old piglets (n=25) underwent asphyxial cardiac arrest followed by randomization and blinding to CPR with 0.21 (n=10) or 1.0 inspired oxygen (n=10) through 10 minutes post return of spontaneous circulation. Sham was n=5. Survivors received 4 hours of protocolized postarrest care, whereupon brain was obtained for mitochondrial analysis and neuropathology. Groups were compared using Kruskal‐Wallis test, Wilcoxon rank‐sum test, and generalized estimating equations regression models. Both 1.0 and 0.21 groups were similar in systemic hemodynamics and cerebral blood flow, as well as survival (8/10). The 1.0 animals had relative cerebral hyperoxia during CPR and immediately following return of spontaneous circulation (brain tissue oxygen tension, 85% [interquartile range, 72%–120%] baseline in 0.21 animals versus 697% [interquartile range, 515%–721%] baseline in 1.0 animals; P=0.001 at 10 minutes postarrest). Cerebral mitochondrial reactive oxygen species production was higher in animals treated with 1.0 compared with 0.21 (P<0.03). Exposure to 1.0 oxygen led to increased cerebral oxidative injury to proteins and lipids, as evidenced by significantly higher protein carbonyls and 4‐hydroxynoneals compared with 0.21 (P<0.05) and sham (P<0.001). Conclusions Exposure to 1.0 inspired oxygen during CPR caused cerebral hyperoxia during resuscitation, and resultant increased mitochondrial‐derived reactive oxygen species and oxidative injury following cardiac arrest.
Collapse
Affiliation(s)
- Alexandra M Marquez
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Ryan W Morgan
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Tiffany Ko
- Division of Neurology Department of Pediatrics Children's Hospital of Philadelphia PA
| | - William P Landis
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Marco M Hefti
- Department of Pathology University of Iowa Iowa City IA
| | - Constantine D Mavroudis
- Division of Cardiothoracic Surgery Department of Surgery Children's Hospital of Philadelphia PA
| | - Meagan J McManus
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | | | - Jonathan Starr
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Anna L Roberts
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Yuxi Lin
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Vinay Nadkarni
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Daniel J Licht
- Division of Neurology Department of Pediatrics Children's Hospital of Philadelphia PA
| | - Robert A Berg
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Robert M Sutton
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Todd J Kilbaugh
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| |
Collapse
|
85
|
Zimering MB, Pulikeyil AT, Myers CE, Pang KC. Serotonin 2A Receptor Autoantibodies Increase in Adult Traumatic Brain Injury In Association with Neurodegeneration. JOURNAL OF ENDOCRINOLOGY AND DIABETES 2020; 7:1-8. [PMID: 32671201 PMCID: PMC7362960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
OBJECTIVE Traumatic brain injury (TBI) is associated with an increased risk of late neurodegenerative complications via unknown mechanisms. Circulating neurotoxic 5-hydroxytryptamine 2A receptor (5-HT2AR) autoantibodies were reported to increase in subsets of obese type 2 diabetes having microvascular complications. We tested whether 5-HT2AR autoantibodies increase in adults following traumatic brain injury in association with neurodegenerative complications. METHODS Plasma from thirty-five middle-aged and older adult veterans (mean 65 years old) who had suffered traumatic brain injury was subjected to protein-A affinity chromatography. The resulting immunoglobulin (Ig) G fraction was tested for neurotoxicity (acute neurite retraction, and accelerated cell death) in mouse N2A neuroblastoma cells or for binding to a linear synthetic peptide corresponding to the second extracellular loop region of the human 5-HT2A receptor. RESULTS Nearly two-thirds of traumatic brain injured-patients harbored 5-HT2AR autoantibodies in their circulation. Active TBI autoantibodies caused neurite retraction in mouse N2A neuroblastoma cells and accelerated N2A cell loss which was substantially prevented by co-incubation with a two hundred and fifty nanomolar concentration of M100907, a highly selective 5-HT2AR antagonist. Antagonists of RhoA/Rho kinase and Gq11/phospholipase C/inositol triphosphate receptor signaling pathways blocked TBI autoantibody-induced neurite retraction. Following traumatic brain injury, autoantibody binding to a 5-HT2A receptor peptide was significantly increased in patients having co-morbid Parkinson's disease (n=3), dementia (n=5), and painful neuropathy (n=8) compared to TBI subsets without neurologic or microvascular complication (n=20). Autoantibody titer was significantly elevated in TBI subsets experiencing multiple neurotraumatic exposures vs. single TBI. Plasma white blood cell, a marker of systemic inflammation, correlated significantly (correlation coefficient r =0.52; P < 0.01) with, 5-HT2A receptor peptide binding of the TBI-autoantibody. CONCLUSION These data suggest that circulating neurotoxic 5-hydroxytryptamine 2A receptor agonist autoantibodies increase in adults following traumatic brain injury in association with late neurodegenerative complications.
Collapse
Affiliation(s)
- Mark B. Zimering
- Veterans Affairs New Jersey Healthcare System, East Orange, NJ
- Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ
| | | | - Catherine E. Myers
- Veterans Affairs New Jersey Healthcare System, East Orange, NJ
- Rutgers-New Jersey Medical School, Newark, NJ
| | - Kevin C. Pang
- Veterans Affairs New Jersey Healthcare System, East Orange, NJ
- Rutgers-New Jersey Medical School, Newark, NJ
| |
Collapse
|
86
|
Zhang ZW, Liang J, Yan JX, Ye YC, Wang JJ, Chen C, Sun HT, Chen F, Tu Y, Li XH. TBHQ improved neurological recovery after traumatic brain injury by inhibiting the overactivation of astrocytes. Brain Res 2020; 1739:146818. [PMID: 32275911 DOI: 10.1016/j.brainres.2020.146818] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) is a major leading cause of death and long-term disability. Although astrocytes play a key role in neuroprotection after TBI in the early stage, the overactivation of astrocytes can lead to long-term functional deficits, and the underlying pathophysiological mechanisms remain unclear. In addition, it is unknown whether the nuclear factor erythroid 2-related factor2/haem oxygenase-1 (Nrf-2/HO-1) pathway could elicit a neuroprotective effect by decreasing astrocyte overactivation after TBI. We aimed to study the effects of tert-butylhydroquinone (TBHQ) in reducing astrocyte overactivation after TBI and explored the underlying mechanisms. We first established a controlled cortical impact (CCI) model in rats and performed Haematoxylin and eosin (H&E) staining to observe brain tissue damage. The cognitive function of rats was assessed by modified neurological severity scoring (mNSS) and Morris water maze (MWM) test. Astrocyte and microglia activation was detected by immunofluorescence staining. Oxidative stress conditions were investigated using Western blotting. An enzyme-linked immunosorbent assay (ELISA) was designed to assess the level of the proinflammatory factor tumour necrosis factor-alpha (TNF-α). Dihydroethidium (DHE) staining was used to detect reactive oxygen species (ROS). Apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The results showed that the administration of TBHQ ameliorated motor function and cognitive deficits and decreased the lesion volume. In addition, TBHQ significantly decreased astrocyte overactivation, diminished the pro-inflammatory phenotype M1 and inflammatory cytokines production after TBI, increased Nrf-2 nuclear accumulation, and enhanced the levels of the Nrf-2 downstream antioxidative genes HO-1 and NADPH-quinone oxidoreductase-1 (NQO-1). Furthermore, TBHQ treatment alleviated apoptosis and neuronal death in the cerebral cortex. Overall, our data indicated that the upregulation of Nrf-2 expression could enhance neuroprotection and decrease astrocyte overactivation and might represent a new theoretical basis for treating TBI.
Collapse
Affiliation(s)
- Zhen-Wen Zhang
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jun Liang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jing-Xing Yan
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Yi-Chao Ye
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Chong Chen
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Feng Chen
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yue Tu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China.
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
87
|
Honig MG, Dorian CC, Worthen JD, Micetich AC, Mulder IA, Sanchez KB, Pierce WF, Del Mar NA, Reiner A. Progressive long-term spatial memory loss following repeat concussive and subconcussive brain injury in mice, associated with dorsal hippocampal neuron loss, microglial phenotype shift, and vascular abnormalities. Eur J Neurosci 2020; 54:5844-5879. [PMID: 32090401 PMCID: PMC7483557 DOI: 10.1111/ejn.14711] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/14/2022]
Abstract
There is considerable concern about the long‐term deleterious effects of repeat head trauma on cognition, but little is known about underlying mechanisms and pathology. To examine this, we delivered four air blasts to the left side of the mouse cranium, a week apart, with an intensity that causes deficits when delivered singly and considered “concussive,” or an intensity that does not yield significant deficits when delivered singly and considered “subconcussive.” Neither repeat concussive nor subconcussive blast produced spatial memory deficits at 4 months, but both yielded deficits at 14 months, and dorsal hippocampal neuron loss. Hierarchical cluster analysis of dorsal hippocampal microglia across the three groups based on morphology and expression of MHCII, CX3CR1, CD68 and IBA1 revealed five distinct phenotypes. Types 1A and 1B microglia were more common in sham mice, linked to better neuron survival and memory, and appeared mildly activated. By contrast, 2B and 2C microglia were more common in repeat concussive and subconcussive mice, linked to poorer neuron survival and memory, and characterized by low expression levels and attenuated processes, suggesting they were de‐activated and dysfunctional. In addition, endothelial cells in repeat concussive mice exhibited reduced CD31 and eNOS expression, which was correlated with the prevalence of type 2B and 2C microglia. Our findings suggest that both repeat concussive and subconcussive head injury engender progressive pathogenic processes, possibly through sustained effects on microglia that over time lead to increased prevalence of dysfunctional microglia, adversely affecting neurons and blood vessels, and thereby driving neurodegeneration and memory decline.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Conor C Dorian
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - John D Worthen
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anthony C Micetich
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Isabelle A Mulder
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Katelyn B Sanchez
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - William F Pierce
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
88
|
Staniloiu A, Kordon A, Markowitsch HJ. Stress- and trauma-related blockade of episodic-autobiographical memory processing. Neuropsychologia 2020; 139:107364. [PMID: 32006541 DOI: 10.1016/j.neuropsychologia.2020.107364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 01/14/2020] [Accepted: 01/24/2020] [Indexed: 12/21/2022]
Abstract
Memory disorders without a direct neural substrate still belong to the riddles in neuroscience. Although they were for a while dissociated from research and clinical arenas, risking becoming forgotten diseases, they sparked novel interests, paralleling the refinements in functional neuroimaging and neuropsychology. Although Endel Tulving has not fully embarked himself on exploring this field, he had published at least one article on functional amnesia (Schacter et al., 1982) and ignited a seminal article on amnesia with mixed etiology (Craver et al., 2014). Most importantly, the research of Endel Tulving has provided the researchers and clinicians in the field of dissociative or functional amnesia with the best framework for superiorly understanding these disorders through the lens of his evolving concept of episodic memory and five long term memory systems classification, which he developed and advanced. Herein we use the classification of long-term memory systems of Endel Tulving as well as his concepts and views on autonoetic consciousness, relationships between memory systems and relationship between episodic memory and emotion to describe six cases of dissociative amnesia that put a challenge for researchers and clinicians due to their atypicality. We then discuss their possible triggering and maintaining mechanisms, pointing to their clinical heterogeneity and multifaceted causally explanatory frameworks.
Collapse
Affiliation(s)
- Angelica Staniloiu
- University of Bielefeld, Germany; University of Bucharest, Romania; Oberberg Clinic Hornberg, Germany
| | - Andreas Kordon
- Oberberg Clinic Hornberg, Germany; University of Freiburg, Germany
| | | |
Collapse
|
89
|
Edwards KA, Motamedi V, Osier ND, Kim HS, Yun S, Cho YE, Lai C, Dell KC, Carr W, Walker P, Ahlers S, LoPresti M, Yarnell A, Tschiffley A, Gill JM. A Moderate Blast Exposure Results in Dysregulated Gene Network Activity Related to Cell Death, Survival, Structure, and Metabolism. Front Neurol 2020; 11:91. [PMID: 32174881 PMCID: PMC7054450 DOI: 10.3389/fneur.2020.00091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 01/27/2020] [Indexed: 12/11/2022] Open
Abstract
Blast exposure is common in military personnel during training and combat operations, yet biological mechanisms related to cell survival and function that coordinate recovery remain poorly understood. This study explored how moderate blast exposure influences gene expression; specifically, gene-network changes following moderate blast exposure. On day 1 (baseline) of a 10-day military training program, blood samples were drawn, and health and demographic information collected. Helmets equipped with bilateral sensors worn throughout training measured overpressure in pounds per square inch (psi). On day 7, some participants experienced moderate blast exposure (peak pressure ≥5 psi). On day 10, 3 days post-exposure, blood was collected and compared to baseline with RNA-sequencing to establish gene expression changes. Based on dysregulation data from RNA-sequencing, followed by top gene networks identified with Ingenuity Pathway Analysis, a subset of genes was validated (NanoString). Five gene networks were dysregulated; specifically, two highly significant networks: (1) Cell Death and Survival (score: 42), including 70 genes, with 50 downregulated and (2) Cell Structure, Function, and Metabolism (score: 41), including 69 genes, with 41 downregulated. Genes related to ubiquitination, including neuronal development and repair: UPF1, RNA Helicase and ATPase (UPF1) was upregulated while UPF3 Regulator of Nonsense Transcripts Homolog B (UPF3B) was downregulated. Genes related to inflammation were upregulated, including AKT serine/threonine kinase 1 (AKT1), a gene coordinating cellular recovery following TBIs. Moderate blast exposure induced significant gene expression changes including gene networks involved in (1) cell death and survival and (2) cellular development and function. The present findings may have implications for understanding blast exposure pathology and subsequent recovery efforts.
Collapse
Affiliation(s)
- Katie A Edwards
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States.,The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Vida Motamedi
- Wake Forest School of Medicine, Wake Forest University, Winston-Salem, NC, United States
| | - Nicole D Osier
- School of Nursing, University of Texas at Austin, Austin, TX, United States.,Department of Neurology, University of Texas, Austin, TX, United States
| | - Hyung-Suk Kim
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Sijung Yun
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Young-Eun Cho
- College of Nursing, University of Iowa, Iowa City, IA, United States
| | - Chen Lai
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Kristine C Dell
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Walter Carr
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Peter Walker
- Naval Medical Research Center, Silver Spring, MD, United States
| | - Stephen Ahlers
- Naval Medical Research Center, Silver Spring, MD, United States
| | - Matthew LoPresti
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Angela Yarnell
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Anna Tschiffley
- Naval Medical Research Center, Silver Spring, MD, United States
| | - Jessica M Gill
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States.,CNRM Co-Director Biomarkers Core, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
90
|
Hartnell D, Gillespie-Jones K, Ciornei C, Hollings A, Thomas A, Harrild E, Reinhardt J, Paterson DJ, Alwis D, Rajan R, Hackett MJ. Characterization of Ionic and Lipid Gradients within Corpus Callosum White Matter after Diffuse Traumatic Brain Injury in the Rat. ACS Chem Neurosci 2020; 11:248-257. [PMID: 31850738 DOI: 10.1021/acschemneuro.9b00257] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There is increased recognition of the effects of diffuse traumatic brain injury (dTBI), which can initiate yet unknown biochemical cascades, resulting in delayed secondary brain degeneration and long-term neurological sequela. There is limited availability of therapies that minimize the effect of secondary brain damage on the quality of life of people who have suffered TBI, many of which were otherwise healthy adults. Understanding the cascade of biochemical events initiated in specific brain regions in the acute phase of dTBI and how this spreads into adjacent brain structures may provide the necessary insight into drive development of improved therapies. In this study, we have used direct biochemical imaging techniques (Fourier transform infrared spectroscopic imaging) and elemental mapping (X-ray fluorescence microscopy) to characterize biochemical and elemental alterations that occur in corpus callosum white matter in the acute phase of dTBI. The results provide direct visualization of differential biochemical and ionic changes that occur in the highly vulnerable medial corpus callosum white matter relative to the less vulnerable lateral regions of the corpus callosum. Specifically, the results suggest that altered ionic gradients manifest within mechanically damaged medial corpus callosum, potentially spreading to and inducing lipid alterations to white matter structures in lateral brain regions.
Collapse
Affiliation(s)
- David Hartnell
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Sciences, Curtin University, Perth, Western Australia, Australia 6845
- Curtin Health Innovation Research Institute, Curtin University, Perth, AUS 6102
| | - Kate Gillespie-Jones
- Neuroscience Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia 3168
| | - Cristina Ciornei
- Neuroscience Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia 3168
| | - Ashley Hollings
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Sciences, Curtin University, Perth, Western Australia, Australia 6845
- Curtin Health Innovation Research Institute, Curtin University, Perth, AUS 6102
| | - Alexander Thomas
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Sciences, Curtin University, Perth, Western Australia, Australia 6845
- Curtin Health Innovation Research Institute, Curtin University, Perth, AUS 6102
| | - Elizabeth Harrild
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Sciences, Curtin University, Perth, Western Australia, Australia 6845
- Curtin Health Innovation Research Institute, Curtin University, Perth, AUS 6102
| | - Juliane Reinhardt
- Australian Nuclear Science and Technology Organisation, 800 Blackburn Road, Clayton, Victoria, Australia 3168
- Department of Chemistry and Physics, ARC Centre of Excellence for Advanced Molecular Imaging, Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, Australia 3086
| | - David J. Paterson
- Australian Nuclear Science and Technology Organisation, 800 Blackburn Road, Clayton, Victoria, Australia 3168
| | - Dasuni Alwis
- Neuroscience Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia 3168
| | - Ramesh Rajan
- Neuroscience Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia 3168
| | - Mark J. Hackett
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Sciences, Curtin University, Perth, Western Australia, Australia 6845
- Curtin Health Innovation Research Institute, Curtin University, Perth, AUS 6102
| |
Collapse
|
91
|
Ren YZ, Zhang BZ, Zhao XJ, Zhang ZY. Resolvin D1 ameliorates cognitive impairment following traumatic brain injury via protecting astrocytic mitochondria. J Neurochem 2020; 154:530-546. [PMID: 31951012 DOI: 10.1111/jnc.14962] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/29/2019] [Accepted: 01/02/2020] [Indexed: 12/18/2022]
Abstract
Cognitive impairment is one of the most common and devastating neuropsychiatric sequelae after traumatic brain injury (TBI), and hippocampal neuronal survival plays a causal role in this pathological process. Resolvin D1 (RvD1), an important endogenous specialized pro-resolving mediator, has recently been reported to exert a potent protective effect on mitochondria. This suggests that RvD1 may suppress neuroinflammation and protect astrocytic mitochondria at the same time to play further neuroprotective roles. C57BL/6 mice subjected to TBI using a controlled cortical impact device were used for in vivo experiments. Cultured primary mouse astrocytes and an N2a mouse neuroblastoma cell line were used for in vitro experiments. In TBI mice, RvD1 significantly ameliorated cognitive impairment, suppressed gliosis and alleviated neuronal loss in the hippocampus. To explore the mechanism underlying this activity, we verified that RvD1 can induce a higher level of mitophagy to remove damaged mitochondria and eliminate extra mitochondria-derived reactive oxygen species (mitoROS) by activating ALX4/FPR2 receptors in astrocytes. In an in vitro model, we further confirmed that RvD1 can protect mitochondrial morphology and membrane potential in astrocytes and thereby enhance the survival of neurons. Meanwhile, RvD1 was also shown to increase the expression of brain-derived neurotrophic factor and glutamate aspartate transporter in the hippocampus following TBI, which indicates a possible way by which RvD1 increases the supportive function of astrocytes. These findings suggest that RvD1 may be a potent therapeutic option for ameliorating cognitive impairment following TBI by controlling neuroinflammation and protecting astrocytic mitochondria.
Collapse
Affiliation(s)
- Yi-Zhi Ren
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Ben-Zheng Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xiao-Jing Zhao
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Zhi-Yuan Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, China.,The Key Laboratory of Antibody Technique of the Ministry of Health, Nanjing Medical University, Nanjing, China.,Department of Neurology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
92
|
Greig NH, Lecca D, Hsueh SC, Nogueras-Ortiz C, Kapogiannis D, Tweedie D, Glotfelty EJ, Becker RE, Chiang YH, Hoffer BJ. (-)-Phenserine tartrate (PhenT) as a treatment for traumatic brain injury. CNS Neurosci Ther 2019; 26:636-649. [PMID: 31828969 PMCID: PMC7248544 DOI: 10.1111/cns.13274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/14/2019] [Accepted: 11/16/2019] [Indexed: 12/21/2022] Open
Abstract
Aim Traumatic brain injury (TBI) is one of the most common causes of morbidity and mortality of both young adults and the elderly, and is a key contributing factor in about 30% of all injury‐associated deaths occurring within the United States of America. Albeit substantial impact has been made to improve our comprehension of the mechanisms that underpin the primary and secondary injury stages initiated by a TBI incident, this knowledge has yet to successfully translate into the development of an effective TBI pharmacological treatment. Developing consent suggests that a TBI can concomitantly trigger multiple TBI‐linked cascades that then progress in parallel and, if correct, the multifactorial nature of TBI would make the discovery of a single effective mechanism‐targeted drug unlikely. Discussion We review recent data indicating that the small molecular weight drug (−)‐phenserine tartrate (PhenT), originally developed for Alzheimer's disease (AD), effectively inhibits a broad range of mechanisms pertinent to mild (m) and moderate (mod)TBI, which in combination underpin the ensuing cognitive and motor impairments. In cellular and animal models at clinically translatable doses, PhenT mitigated mTBI‐ and modTBI‐induced programmed neuronal cell death (PNCD), oxidative stress, glutamate excitotoxicity, neuroinflammation, and effectively reversed injury‐induced gene pathways leading to chronic neurodegeneration. In addition to proving efficacious in well‐characterized animal TBI models, significantly mitigating cognitive and motor impairments, the drug also has demonstrated neuroprotective actions against ischemic stroke and the organophosphorus nerve agent and chemical weapon, soman. Conclusion In the light of its tolerability in AD clinical trials, PhenT is an agent that can be fast‐tracked for evaluation in not only civilian TBI, but also as a potentially protective agent in battlefield conditions where TBI and chemical weapon exposure are increasingly jointly occurring.
Collapse
Affiliation(s)
- Nigel H Greig
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Daniela Lecca
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Shih-Chang Hsueh
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.,The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Carlos Nogueras-Ortiz
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - David Tweedie
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Elliot J Glotfelty
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Robert E Becker
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.,Aristea Translational Medicine Corporation, Park City, UT, USA
| | - Yung-Hsiao Chiang
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Barry J Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
93
|
Ongnok B, Chattipakorn N, Chattipakorn SC. Doxorubicin and cisplatin induced cognitive impairment: The possible mechanisms and interventions. Exp Neurol 2019; 324:113118. [PMID: 31756316 DOI: 10.1016/j.expneurol.2019.113118] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/18/2019] [Accepted: 11/15/2019] [Indexed: 12/24/2022]
Abstract
Chemotherapy has significantly increased the number of cancer survivors. However, chemotherapy itself carries various adverse effects that limit the efficacy of treatment and quality of life of the cancer patients. Most patients who have received chemotherapy report some cognitive deficit characterized by dysfunction in memory, learning, concentration, and reasoning. The phenomenon of cognitive decline developed from chemotherapy treatment is referred to as chemotherapy-induced cognitive impairment (CICI) or chemobrain. The two most common cancers occurring worldwide are lung and breast cancer. The predominant chemotherapeutic drugs used to treat lung and breast cancer are doxorubicin and cisplatin. There is evidence to suggest that both drugs potentially induce chemobrain. The evidence around the proposed pathogenesis of chemobrain caused by these two drugs is inconsistent. Understanding the underlying mechanisms involved in the development of chemobrain would aid in the prevention or treatment of the adverse effects of chemotherapy on brain. This review will summarize and discuss controversial findings and possible mechanisms involved in the development of chemobrain and the interventions which could limit it from in vitro, in vivo, and clinical studies.
Collapse
Affiliation(s)
- Benjamin Ongnok
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
94
|
Chien YJ, Chien YC, Liu CT, Wu HC, Chang CY, Wu MY. Effects of Methylphenidate on Cognitive Function in Adults with Traumatic Brain Injury: A Meta-Analysis. Brain Sci 2019; 9:brainsci9110291. [PMID: 31653039 PMCID: PMC6895997 DOI: 10.3390/brainsci9110291] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 12/29/2022] Open
Abstract
This meta-analysis evaluated the effects of methylphenidate (MPH) on cognitive outcome and adverse events in adults with traumatic brain injuries (TBI). We searched PubMed, EMBASE, and PsycINFO for randomized controlled trials (RCTs) published before July 2019. Studies that compared the effects of MPH and placebos in adults with TBI were included. The primary outcome was cognitive function, while the secondary outcome was adverse events. Meta-regression and sensitivity analysis were conducted to evaluate heterogeneity. Seventeen RCTs were included for qualitative analysis, and ten RCTs were included for quantitative analysis. MPH significantly improved processing speed, measured by Choice Reaction Time (standardized mean difference (SMD): -0.806; 95% confidence interval (CI): -429 to -0.182, p = 0.011) and Digit Symbol Coding Test (SMD: -0.653; 95% CI: -1.016 to -0.289, p < 0.001). Meta-regression showed that the reaction time was inversely associated with the duration of MPH. MPH administration significantly increased heart rate (SMD: 0.553; 95% CI: 0.337 to 0.769, p < 0.001), while systolic or diastolic blood pressure did not exhibit significant differences. Therefore, MPH elicited better processing speed in adults with TBI. However, MPH use could significantly increase heart rate. A larger study is required to evaluate the effect of dosage, age, or optimal timing on treatment of adults with TBI.
Collapse
Affiliation(s)
- Yung-Jiun Chien
- Department of Physical Medicine and Rehabilitation, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 231 New Taipei, Taiwan.
- School of Medicine, Tzu Chi University, 970 Hualien, Taiwan.
| | - Yung-Chen Chien
- Department of Medical Education, Taipei Medical University Hospital, 110 Taipei, Taiwan.
| | - Chien-Ting Liu
- Department of Physical Medicine and Rehabilitation, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 231 New Taipei, Taiwan.
- School of Medicine, Tzu Chi University, 970 Hualien, Taiwan.
| | - Hsin-Chi Wu
- Department of Physical Medicine and Rehabilitation, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 231 New Taipei, Taiwan.
- School of Medicine, Tzu Chi University, 970 Hualien, Taiwan.
| | - Chun-Yu Chang
- School of Medicine, Tzu Chi University, 970 Hualien, Taiwan.
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 231 New Taipei, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, 970 Hualien, Taiwan.
| |
Collapse
|
95
|
Bugay V, Bozdemir E, Vigil FA, Chun SH, Holstein DM, Elliott WR, Sprague CJ, Cavazos JE, Zamora DO, Rule G, Shapiro MS, Lechleiter JD, Brenner R. A Mouse Model of Repetitive Blast Traumatic Brain Injury Reveals Post-Trauma Seizures and Increased Neuronal Excitability. J Neurotrauma 2019; 37:248-261. [PMID: 31025597 DOI: 10.1089/neu.2018.6333] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Repetitive blast traumatic brain injury (TBI) affects numerous soldiers on the battlefield. Mild TBI has been shown to have long-lasting effects with repeated injury. We have investigated effects on neuronal excitability after repetitive, mild TBI in a mouse model of blast-induced brain injury. We exposed mice to mild blast trauma of an average peak overpressure of 14.6 psi, repeated across three consecutive days. While a single exposure did not reveal trauma as indicated by the glial fibrillary acidic protein indicator, three repetitive blasts did show significant increases. As well, mice had an increased indicator of inflammation (Iba-1) and increased tau, tau phosphorylation, and altered cytokine levels in the spleen. Video-electroencephalographic monitoring 48 h after the final blast exposure demonstrated seizures in 50% (12/24) of the mice, most of which were non-convulsive seizures. Long-term monitoring revealed that spontaneous seizures developed in at least 46% (6/13) of the mice. Patch clamp recording of dentate gyrus hippocampus neurons 48 h post-blast TBI demonstrated a shortened latency to the first spike and hyperpolarization of action potential threshold. We also found that evoked excitatory postsynaptic current amplitudes were significantly increased. These findings indicate that mild, repetitive blast exposures cause increases in neuronal excitability and seizures and eventual epilepsy development in some animals. The non-convulsive nature of the seizures suggests that subclinical seizures may occur in individuals experiencing even mild blast events, if repeated.
Collapse
Affiliation(s)
- Vladislav Bugay
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Eda Bozdemir
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Fabio A Vigil
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Sang H Chun
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Deborah M Holstein
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - William R Elliott
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Cassie J Sprague
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Jose E Cavazos
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas.,Department of Neurology, University of Texas Health San Antonio, San Antonio, Texas
| | - David O Zamora
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | | | - Mark S Shapiro
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Robert Brenner
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
96
|
Tadepalli SA, Bali ZK, Bruszt N, Nagy LV, Amrein K, Fazekas B, Büki A, Czeiter E, Hernádi I. Long-term cognitive impairment without diffuse axonal injury following repetitive mild traumatic brain injury in rats. Behav Brain Res 2019; 378:112268. [PMID: 31580914 DOI: 10.1016/j.bbr.2019.112268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/26/2019] [Accepted: 09/29/2019] [Indexed: 12/30/2022]
Abstract
Repetitive mild traumatic brain injuries (TBI) impair cognitive abilities and increase risk of neurodegenerative disorders in humans. We developed two repetitive mild TBI models in rats with different time intervals between successive weight-drop injuries. Rats were subjected to repetitive Sham (no injury), single mild (mTBI), repetitive mild (rmTBI - 5 hits, 24 h apart), rapid repetitive mild (rapTBI - 5 hits, 5 min apart) or a single severe (sTBI) TBI. Cognitive performance was assessed 2 and 8 weeks after TBI in the novel object recognition test (NOR), and 6-7 weeks after TBI in the water maze (MWM). Acute immunohistochemical markers were evaluated 24 h after TBI, and blood biomarkers were measured with ELISA 8 weeks after TBI. In the NOR, both rmTBI and rapTBI showed poor performance at 2 weeks post-injury. At 8 weeks post-injury, the rmTBI group still performed worse than the Sham and mTBI groups, while the rapTBI group recovered. In the MWM, the rapTBI group performed worse than the Sham and mTBI groups. Acute APP and RMO-14 immunohistochemistry showed axonal injury at the pontomedullary junction in the sTBI, but not in other groups. ELISA showed increased serum GFAP levels 8 weeks after sTBI, while no differences were found between the injury groups in the levels of phosphorylated-tau and S100β. Results suggest that the rmTBI protocol is the most suitable model for testing cognitive impairment after mild repetitive head injuries and that the prolonged cognitive impairment after repetitive mild TBI originates from different structural and molecular mechanisms compared to similar impairments after single sTBI.
Collapse
Affiliation(s)
- Sai Ambika Tadepalli
- Department of Experimental Zoology and Neurobiology, Faculty of Sciences, University of Pécs, Ifjúság útja 6, H-7624 Pécs, Hungary; Translational Neuroscience Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Center for Neuroscience, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Zsolt Kristóf Bali
- Translational Neuroscience Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Center for Neuroscience, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Grastyán Translational Research Centre, University of Pécs, Ifjúság útja 6, H-7624 Pécs, Hungary.
| | - Nóra Bruszt
- Translational Neuroscience Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Center for Neuroscience, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Institute of Physiology, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs Hungary
| | - Lili Veronika Nagy
- Department of Experimental Zoology and Neurobiology, Faculty of Sciences, University of Pécs, Ifjúság útja 6, H-7624 Pécs, Hungary; Translational Neuroscience Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Center for Neuroscience, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Krisztina Amrein
- Center for Neuroscience, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Department of Neurosurgery, Medical School, University of Pécs, Rét u. 2, H-7623 Pécs, Hungary; Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Bálint Fazekas
- Center for Neuroscience, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Department of Neurosurgery, Medical School, University of Pécs, Rét u. 2, H-7623 Pécs, Hungary; Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - András Büki
- Center for Neuroscience, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Department of Neurosurgery, Medical School, University of Pécs, Rét u. 2, H-7623 Pécs, Hungary; Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Endre Czeiter
- Center for Neuroscience, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Department of Neurosurgery, Medical School, University of Pécs, Rét u. 2, H-7623 Pécs, Hungary; Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; MTA-PTE Clinical Neuroscience MR Research Group, Rét u. 2, H-7623 Pécs, Hungary
| | - István Hernádi
- Department of Experimental Zoology and Neurobiology, Faculty of Sciences, University of Pécs, Ifjúság útja 6, H-7624 Pécs, Hungary; Translational Neuroscience Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Center for Neuroscience, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Grastyán Translational Research Centre, University of Pécs, Ifjúság útja 6, H-7624 Pécs, Hungary; Institute of Physiology, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs Hungary
| |
Collapse
|
97
|
Charkviani M, Muradashvili N, Lominadze D. Vascular and non-vascular contributors to memory reduction during traumatic brain injury. Eur J Neurosci 2019; 50:2860-2876. [PMID: 30793398 PMCID: PMC6703968 DOI: 10.1111/ejn.14390] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/06/2019] [Accepted: 02/07/2019] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) is an increasing health problem. It is a complex, progressive disease that consists of many factors affecting memory. Studies have shown that increased blood-brain barrier (BBB) permeability initiates pathological changes in neuro-vascular network but the role of cerebrovascular dysfunction and its mediated mechanisms associated with memory reduction during TBI are still not well understood. Changes in BBB, inflammation, extravasation of blood plasma components, activation of neuroglia lead to neurodegeneration. Extravasated proteins such as amyloid-beta, fibrinogen, and cellular prion protein may form degradation resistant complexes that can lead to neuronal dysfunction and degeneration. They also have the ability to activate astrocytes, and thus, can be involved in memory impairment. Understanding the triggering mechanisms and the places they originate in vasculature or in extravascular tissue may help to identify potential therapeutic targets to ameliorate memory reduction during TBI. The goal of this review is to discuss conceptual mechanisms that lead to short-term memory reduction during non-severe TBI considering distinction between vascular and non-vascular effects on neurons. Some aspects of these mechanisms need to be confirmed further. Therefore, we hope that the discussion presented bellow may lead to experiments that may clarify the triggering mechanisms of memory reduction after head trauma.
Collapse
Affiliation(s)
- Mariam Charkviani
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
- Department of Basic Medicine, Caucasus International University, Tbilisi, Georgia
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
- Kentucky Spinal Cord Research Center, University of Louisville, School of Medicine, Louisville, KY, USA
| |
Collapse
|
98
|
Cassol G, Godinho DB, de Zorzi VN, Farinha JB, Della-Pace ID, de Carvalho Gonçalves M, Oliveira MS, Furian AF, Fighera MR, Royes LFF. Potential therapeutic implications of ergogenic compounds on pathophysiology induced by traumatic brain injury: A narrative review. Life Sci 2019; 233:116684. [DOI: 10.1016/j.lfs.2019.116684] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/22/2019] [Indexed: 12/19/2022]
|
99
|
Edwards G, Zhao J, Dash PK, Soto C, Moreno-Gonzalez I. Traumatic Brain Injury Induces Tau Aggregation and Spreading. J Neurotrauma 2019; 37:80-92. [PMID: 31317824 PMCID: PMC6921297 DOI: 10.1089/neu.2018.6348] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The misfolding and aggregation of tau protein into neurofibrillary tangles is the main underlying hallmark of tauopathies. Most tauopathies have a sporadic origin and can be associated with multiple risk factors. Traumatic brain injury (TBI) has been suggested as a risk factor for tauopathies by triggering disease onset and facilitating its progression. Several studies indicate that TBI seems to be a risk factor to development of Alzheimer disease and chronic traumatic encephalopathy, because there is a relationship of TBI severity and propensity to development of these illnesses. In this study, we evaluated whether moderate to severe TBI can trigger the initial formation of pathological tau that would induce the development of the pathology throughout the brain. To this end, we subjected tau transgenic mice to TBI and assessed tau phosphorylation and aggregation pattern to create a spatial heat map of tau deposition and spreading in the brain. Our results suggest that brain injured tau transgenic mice have an accelerated tau pathology in different brain regions that increases over time compared with sham mice. The appearance of pathological tau occurs in regions distant to the injury area that are connected synaptically, suggesting dissemination of tau aggregates. Overall, this work posits TBI as a risk factor for tauopathies through the induction of tau hyperphosphorylation and aggregation.
Collapse
Affiliation(s)
- George Edwards
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Jing Zhao
- Department of Neurobiology and Anatomy, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Ines Moreno-Gonzalez
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, Texas.,Department of Cell Biology, Networking Research Center on Neurodegenerative Diseases (CIBERNED), Facultad Ciencias, Universidad de Malaga, Malaga, Spain
| |
Collapse
|
100
|
El-Gazar AA, Soubh AA, Mohamed EA, Awad AS, El-Abhar HS. Morin post-treatment confers neuroprotection in a novel rat model of mild repetitive traumatic brain injury by targeting dementia markers, APOE, autophagy and Wnt/β-catenin signaling pathway. Brain Res 2019; 1717:104-116. [DOI: 10.1016/j.brainres.2019.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/19/2019] [Accepted: 04/03/2019] [Indexed: 02/07/2023]
|